1
|
Yang D, Peng D, Zhou Y, Qiang Z, Wan L, Fan X, Meng Y, Xu G, Meng Y. Alpha-Momorcharin, a type I ribosome inactivating protein, induced apoptosis of hepatocellular carcinoma SK-HEP-1 cells through mitochondrial pathway. Nat Prod Res 2025; 39:1128-1138. [PMID: 38126176 DOI: 10.1080/14786419.2023.2295915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023]
Abstract
Alpha-Momorcharin (α-MMC), as one of the most important type I RIPs, has been reported to exert inhibitory effects against various tumour cells through its N-glycosidase activity. The present study was designed to propose an efficient purification strategy and explored its mechanism of apoptosis signalling pathway against human liver cancer cells SK-Hep-1. α-MMC can be successfully obtained by our purification strategy combining ion-exchange and gel-filtration chromatography. The functional studies revealed that α-MMC obviously increased the level of ROS and apoptosis rate, induced cell cycle arrest in the G1 phase, and depolarised MMP of SK-Hep-1 cells. To further confirm whether α-MMC could induce mitochondria involved apoptosis, we found that PARP-1, Caspase-3, Caspase-9, and BCL-2 were downregulated upon α-MMC. Taken together, these results suggested that this natural purified α-MMC can induce apoptosis involved mitochondria and may serve as a potential novel therapeutic drug in the treatment of human liver cancer in the future.
Collapse
Affiliation(s)
- Di Yang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Di Peng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Yiping Zhou
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Sichuan Provincial People's Hospital Jinniu Hospital, Chengdu, Sichuan, China
| | - Zihao Qiang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Li Wan
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiang Fan
- Key Laboratory of Bio-resources and Eco-environment Ministry of Education/Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Yanfa Meng
- Key Laboratory of Bio-resources and Eco-environment Ministry of Education/Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Ge Xu
- The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, Sichuan, China
| | - Yao Meng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Liu Y, Yixilamu, Jin G, Feng M, Chunhua, Dawa. Tibetan golden acupuncture inhibits JNK/caspase-3 signaling pathway to alleviate neuronal apoptosis in cerebral ischemia-reperfusion injury. Heliyon 2024; 10:e40443. [PMID: 39698073 PMCID: PMC11652837 DOI: 10.1016/j.heliyon.2024.e40443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/17/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Background Apoptosis induced by cerebral ischemia-reperfusion is one of the key pathological processes of nerve injury. Tibetan golden acupuncture (GA) is a common treatment for ischemic brain injury in Tibetan. The aim of this study was to explore whether GA prevents cerebral ischemia-reperfusion-induced apoptosis in mice by blocking the JNK/caspase-3 pathway. Methods In experiment I, 36 mice were randomly divided into a Sham group, CI/RI group, CI/RI + GA group. Morris water maze tests, TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining and flow cytometry (FCM) were used to evaluate the effect of the GA intervention on CI/RI. In experiment II, 30 mice were randomly divided into a Sham group, CI/RI group, CI/RI + GA group, CI/RI + SP group and CI/RI + SP + EA group. Western blotting was used to detect protein expression of key factors in the JNK signaling pathway in the hippocampus. Results After 7 and 14 interventions, behavioral evaluations in CI/RI + GA group was significantly different from those in CI/RI groups (p < 0.01), pathological injury and apoptosis were significantly reduced (p < 0.01). Compared with CI/RI group, the expression of P-JNK/JNK, Cleaved caspase-3/caspase-3, Bax, and Bad proteins in CI/RI + GA group, CI/RI + SP and CI/RI + SP + GA groups were significantly decreased (p < 0.01). The expression of B-cell lymphoma 2 (Bcl-2) was significantly increased (p < 0.01, p < 0.05). Conclusions GA can restore neurological dysfunction and inhibit hippocampal neuronal apoptosis in CI/RI mice, at least partially through inhibition of the JNK/Caspase-3 signaling pathway and regulation of apoptosis signals.
Collapse
Affiliation(s)
- Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
- University of Tibetan Medicine, Lhasa, China
| | - Yixilamu
- University of Tibetan Medicine, Lhasa, China
| | - Guilin Jin
- University of Tibetan Medicine, Lhasa, China
| | - Mingke Feng
- University of Tibetan Medicine, Lhasa, China
| | - Chunhua
- University of Tibetan Medicine, Lhasa, China
| | - Dawa
- University of Tibetan Medicine, Lhasa, China
| |
Collapse
|
3
|
Seyrek K, Espe J, Reiss E, Lavrik IN. The Crosstalk of Apoptotic and Non-Apoptotic Signaling in CD95 System. Cells 2024; 13:1814. [PMID: 39513921 PMCID: PMC11545656 DOI: 10.3390/cells13211814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The mechanisms of CD95 (Fas/APO-1)-mediated extrinsic apoptotic pathway in cancer cells have been extensively studied. The majority of human cells express CD95, but not all these cells can induce extrinsic apoptosis. Accumulating evidence has shown that CD95 is a multifunctional protein, and its stimulation can also elicit non-apoptotic or even survival signals. It has become clear that under certain cellular contexts, due to the various checkpoints, CD95 activation can trigger both apoptotic and non-apoptotic signals. The crosstalk of death and survival signals may occur at different levels of signal transduction. The strength of the CD95 stimulation, initial levels of anti-apoptotic proteins, and posttranslational modifications of the core DISC components have been proposed to be the most important factors in the life/death decisions at CD95. Successful therapeutic targeting of CD95 signaling pathways will require a better understanding of the crosstalk between CD95-induced apoptotic and cell survival pathways. In this review, in order to gain a systematic understanding of the crosstalk between CD95-mediated apoptosis and non-apoptotic signaling, we will discuss these issues in a step-by-step way.
Collapse
Affiliation(s)
| | | | | | - Inna N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany; (K.S.); (J.E.); (E.R.)
| |
Collapse
|
4
|
Zhong G, Wang X, Zhang Q, Zhang X, Fang X, Li S, Pan Y, Ma Y, Wang X, Wan T, Wang Q. Exploring the therapeutic implications of natural compounds modulating apoptosis in vascular dementia. Phytother Res 2024; 38:5270-5289. [PMID: 39223915 DOI: 10.1002/ptr.8316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Vascular dementia (VaD) is a prevalent form of dementia stemming from cerebrovascular disease, manifesting in memory impairment and executive dysfunction, thereby imposing a substantial societal burden. Unfortunately, no drugs have been approved for the treatment of VaD due to its intricate pathogenesis, and the development of innovative and efficacious medications is urgently needed. Apoptosis, a programmed cell death process crucial for eliminating damaged or unwanted cells within an organism, assumes pivotal roles in embryonic development and tissue homeostasis maintenance. An increasing body of evidence indicates that apoptosis may significantly influence the onset and progression of VaD, and numerous natural compounds have demonstrated significant therapeutic potential. Here, we discuss the molecular mechanisms underlying apoptosis and its correlation with VaD. We also provide a crucial reference for developing innovative pharmaceuticals by systematically reviewing the latest research progress concerning the neuroprotective effects of natural compounds on VaD by regulating apoptosis. Further high-quality clinical studies are imperative to firmly ascertain these natural compounds' clinical efficacy and safety profiles in the treatment of VaD.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- Department of Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xueying Zhang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China
| | - Xiaoling Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuting Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaru Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujie Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuejing Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Wan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Villaorduña C, Barrios-Arpi L, Lira-Mejía B, Ramos-Gonzalez M, Ramos-Coaguila O, Inostroza-Ruiz L, Romero A, Rodríguez JL. The Fungicide Ipconazole Can Activate Mediators of Cellular Damage in Rat Brain Regions. TOXICS 2024; 12:638. [PMID: 39330566 PMCID: PMC11435560 DOI: 10.3390/toxics12090638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
This study aimed to investigate the toxicity of the fungicide ipconazole on oxidative status, cell death and inflammasome complex activation in the hypothalamus, cerebral cortex, striatum and hippocampus of rats. Female albino rats were randomly divided into a control group and four groups treated with ipconazole at doses of 1, 5, 10 and 20 mg/kg b.w., administered for six days. Ipconazole significantly increased MDA and ROS levels in all brain regions studied, while reducing catalase enzyme activity. The molecular expression of cell death-related genes (AKT1, APAF1, BNIP3, CASP3 and BAX) and the inflammasome complex (CASP1, IL1β, IL6, NLRP3, NFĸB and TNFα) was also assessed, showing increased expression in at least one brain region. The findings demonstrate that ipconazole induces central nervous system toxicity in mammals, highlighting its potential role as a risk factor in the development of neurodegenerative disorders in individuals exposed to this contaminant.
Collapse
Affiliation(s)
- Carlos Villaorduña
- Animal Physiology Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru
| | - Luis Barrios-Arpi
- Animal Physiology Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru
| | - Boris Lira-Mejía
- Animal Physiology Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru
| | - Mariella Ramos-Gonzalez
- Zootecnia an Animal Production Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru
| | - Olger Ramos-Coaguila
- Zootecnia an Animal Production Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru
| | - Luis Inostroza-Ruiz
- Toxicology Laboratory, Faculty of Pharmacy and Biochemistry, Major National University of San Marcos, Lima 15021, Peru
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - José-Luis Rodríguez
- Animal Physiology Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
6
|
Khalef L, Lydia R, Filicia K, Moussa B. Cell viability and cytotoxicity assays: Biochemical elements and cellular compartments. Cell Biochem Funct 2024; 42:e4007. [PMID: 38593323 DOI: 10.1002/cbf.4007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/01/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
Cell viability and cytotoxicity assays play a crucial role in drug screening and evaluating the cytotoxic effects of various chemicals. The quantification of cell viability and proliferation serves as the cornerstone for numerous in vitro assays that assess cellular responses to external factors. In the last decade, several studies have developed guidelines for defining and interpreting cell viability and cytotoxicity based on morphological, biochemical, and functional perspectives. As this domain continues to experience ongoing growth, revealing new mechanisms orchestrating diverse cell cytotoxicity pathways, we suggest a revised classification for multiple assays employed in evaluating cell viability and cell death. This classification is rooted in the cellular compartment and/or biochemical element involved, with a specific focus on mechanistic and essential aspects of the process. The assays are founded on diverse cell functions, encompassing metabolic activity, enzyme activity, cell membrane permeability and integrity, adenosine 5'-triphosphate content, cell adherence, reduction equivalents, dye inclusion or exclusion, constitutive protease activity, colony formation, DNA fragmentation and nuclear splitting. These assays present straightforward, reliable, sensitive, reproducible, cost-effective, and high-throughput approaches for appraising the effects of newly formulated chemotherapeutic biomolecules on the cell survival during the drug development process.
Collapse
Affiliation(s)
- Lefsih Khalef
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Radja Lydia
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Khettar Filicia
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Berkoud Moussa
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| |
Collapse
|
7
|
Tian L, Li Y, Shi Y. Dark and Dronc activation in Drosophila melanogaster. Proc Natl Acad Sci U S A 2024; 121:e2312784121. [PMID: 38381783 PMCID: PMC10907274 DOI: 10.1073/pnas.2312784121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
The onset of apoptosis is characterized by a cascade of caspase activation, where initiator caspases are activated by a multimeric adaptor complex known as the apoptosome. In Drosophila melanogaster, the initiator caspase Dronc undergoes autocatalytic activation in the presence of the Dark apoptosome. Despite rigorous investigations, the activation mechanism for Dronc remains elusive. Here, we report the cryo-EM structures of an auto-inhibited Dark monomer and a single-layered, multimeric Dark/Dronc complex. Our biochemical analysis suggests that the auto-inhibited Dark oligomerizes upon binding to Dronc, which is sufficient for the activation of both Dark and Dronc. In contrast, the previously observed double-ring Dark apoptosome may represent a non-functional or "off-pathway" conformation. These findings expand our understanding on the molecular mechanism of apoptosis in Drosophila.
Collapse
Affiliation(s)
- Lu Tian
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Yini Li
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Yigong Shi
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
- Westlake Laboratory of Life Sciences and Biomedicine, Westlake Institute for Advanced Study, Hangzhou310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou310024, China
| |
Collapse
|
8
|
Liu Y, Yan Z, Ren Y, Wang W, Ke Y, Wang Y, Qi R. Electroacupuncture inhibits hippocampal neuronal apoptosis and improves cognitive dysfunction in mice with vascular dementia via the JNK signaling pathway. Acupunct Med 2023; 41:284-296. [PMID: 36482691 DOI: 10.1177/09645284221136878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
BACKGROUND Electroacupuncture (EA) has been shown to reduce cognitive impairment in vascular dementia (VaD) patients. However, the mechanism of action remains unknown. OBJECTIVE The c-Jun N-terminal kinase (JNK) signaling pathway plays an important role in apoptosis. Herein, we focused on whether EA can inhibit apoptosis and alleviate cognitive impairment by regulating the JNK signaling pathway using a mouse model of VaD induced by modified bilateral common carotid artery occlusion (BCCAo). METHODS In experiment I, 60 mice were randomly divided into a Sham group, BCCAo group, BCCAo + EA group, BCCAo + Sham-EA group, BCCAo + SP group (receiving the selective JNK inhibitor SP600125) and BCCAo + SP + EA group. Morris water maze tests, TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining and flow cytometry were used to evaluate the effect of the EA intervention on VaD. In experiment II, 30 mice were randomly divided into a Sham group, BCCAo group, BCCAo + EA group, BCCAo + SP group and BCCAo + SP + EA group. Western blotting and real-time reverse transcription polymerase chain reaction were used to detect protein and mRNA expression of key factors in the JNK signaling pathway in the hippocampus. RESULTS EA, SP600125 and EA + SP600125 significantly inhibited hippocampal apoptosis and improved cognitive impairment in VaD model mice. There were no significant differences between the BCCAo group and the BCCAo + Sham-EA group. EA, EA + SP600125 and SP600125 inhibited the phosphorylation of JNK and caspase-3. EA and EA + SP600125 promoted protein and mRNA expression of B-cell lymphoma 2 (Bcl-2) in the hippocampus of VaD mice and inhibited protein and mRNA expression of activator protein (AP)-1, p53 and Bax. CONCLUSION EA can reverse cognitive deficits and inhibit hippocampal neuronal apoptosis in VaD model mice, at least partially through inhibition of the JNK signaling pathway and regulation of apoptosis signals.
Collapse
Affiliation(s)
- Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenyang Yan
- Weifang Traditional Chinese Hospital, Weifang, China
| | - Yafei Ren
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Woyu Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yinze Ke
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yifan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Rongming Qi
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Althobaiti F, Sahyon HA, Shanab MMAH, Aldhahrani A, Helal MA, Khireldin A, Shoair AGF, Almalki ASA, Fathy AM. A comparative study of novel ruthenium(III) and iron(III) complexes containing uracil; docking and biological studies. J Inorg Biochem 2023; 247:112308. [PMID: 37441923 DOI: 10.1016/j.jinorgbio.2023.112308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023]
Abstract
Structural and biological studies were conducted on the novel complexes [Fe(U)2(H2O)2]Cl3 (FeU) and [Ru(U)2(H2O)2]Cl3 (RuU) (U = 5,6-Diamino-1,3-dimethylpyrimidine-2,4(1H,3H)-dione) to develop an anticancer drug candidate. The two complexes have been synthesized and characterized. Based on our findings, these complexes have octahedral geometry. The DNA-binding study proved that both complexes coordinated with CT-DNA. The docking study confirmed the potency of both complexes in downregulating the topoisomerase I protein through their high binding affinity. Biological studies have established that both complexes can act as potent anticancer agents against three cancer cell lines. RuU or FeU complexes induce apoptosis in breast cancer cells by increasing caspase9 protein and inhibiting proliferating cell nuclear antigen (PCNA) activity. In addition, both complexes down-regulate topoisomerase I expression in breast cancer cells. Therefore, the RuU and FeU complexes' anticancer activities were mediated via both apoptosis induction and topoisomerase I down-regulation. In conclusion, both complexes have dual anticancer activity pathways that may be responsible for the selective cytotoxicity of the complexes. This makes them more suitable for the development of novel cancer treatment strategies.
Collapse
Affiliation(s)
- Fayez Althobaiti
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Heba A Sahyon
- Chemistry Department, Faculty of Science, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt.
| | - Mai M A H Shanab
- Department of Chemistry, College of Sciences and Humanities Studies (Girls section), Hawtat Bani Tamim 11149, Prince Sattam Bin Abdulaziz University, P.O. Box:13, Saudi Arabia.
| | - Adil Aldhahrani
- Clinical Laboratory Science Department, Turabah University College, Taif University, Taif 21995, Saudi Arabia.
| | - Marihan A Helal
- Chemistry Department, Faculty of Science, Damietta University, Damietta, Egypt
| | - Awad Khireldin
- Air transport management, Singapore Institute of Technology (SIT), Singapore.
| | - Abdel Ghany F Shoair
- Department of Science and Technology, University College-Ranyah, postcode 21975, Taif University, Saudi Arabia; High Altitude Research Center, Taif University, 21944, Saudi Arabia.
| | | | - Ahmed M Fathy
- Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
10
|
Li Y, Tian L, Zhang Y, Shi Y. Structural insights into CED-3 activation. Life Sci Alliance 2023; 6:e202302056. [PMID: 37402593 PMCID: PMC10320015 DOI: 10.26508/lsa.202302056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 07/06/2023] Open
Abstract
In Caenorhabditis elegans (C. elegans), onset of programmed cell death is marked with the activation of CED-3, a process that requires assembly of the CED-4 apoptosome. Activated CED-3 forms a holoenzyme with the CED-4 apoptosome to cleave a wide range of substrates, leading to irreversible cell death. Despite decades of investigations, the underlying mechanism of CED-4-facilitated CED-3 activation remains elusive. Here, we report cryo-EM structures of the CED-4 apoptosome and three distinct CED-4/CED-3 complexes that mimic different activation stages for CED-3. In addition to the previously reported octamer in crystal structures, CED-4, alone or in complex with CED-3, exists in multiple oligomeric states. Supported by biochemical analyses, we show that the conserved CARD-CARD interaction promotes CED-3 activation, and initiation of programmed cell death is regulated by the dynamic organization of the CED-4 apoptosome.
Collapse
Affiliation(s)
- Yini Li
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lu Tian
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Zhang
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yigong Shi
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Westlake Laboratory of Life Science and Biomedicine, Westlake Institute for Advanced Study, Hangzhou, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| |
Collapse
|
11
|
Improvement of Therapeutic Value of Quercetin with Chitosan Nanoparticle Delivery Systems and Potential Applications. Int J Mol Sci 2023; 24:ijms24043293. [PMID: 36834702 PMCID: PMC9959398 DOI: 10.3390/ijms24043293] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
This paper reviews recent studies investigating chitosan nanoparticles as drug delivery systems for quercetin. The therapeutic properties of quercetin include antioxidant, antibacterial and anti-cancer potential, but its therapeutic value is limited by its hydrophobic nature, low bioavailability and fast metabolism. Quercetin may also act synergistically with other stronger drugs for specific disease states. The encapsulation of quercetin in nanoparticles may increase its therapeutic value. Chitosan nanoparticles are a popular candidate in preliminary research, but the complex nature of chitosan makes standardisation difficult. Recent studies have used in-vitro, and in-vivo experiments to study the delivery of quercetin alone or in combination with another active pharmaceutical ingredient encapsulated in chitosan nanoparticles. These studies were compared with the administration of non-encapsulated quercetin formulation. Results suggest that encapsulated nanoparticle formulations are better. In-vivo or animal models simulated the type of disease required to be treated. The types of diseases were breast, lung, liver and colon cancers, mechanical and UVB-induced skin damage, cataracts and general oxidative stress. The reviewed studies included various routes of administration: oral, intravenous and transdermal routes. Although toxicity tests were often included, it is believed that the toxicity of loaded nanoparticles needs to be further researched, especially when not orally administered.
Collapse
|
12
|
Jayathilake AG, Hassanzadeganroudsari M, Jovanovska V, Luwor RB, Nurgali K, Su XQ. The comparative anti-cancer effects of krill oil and oxaliplatin in an orthotopic mouse model of colorectal cancer. Nutr Metab (Lond) 2022; 19:12. [PMID: 35236377 PMCID: PMC8892734 DOI: 10.1186/s12986-022-00646-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 02/09/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Our in vitro studies demonstrated that krill oil (KO) has anti-cancer potential. This study aimed to compare the anti-cancer effects of KO with a commonly used chemotherapeutic drug, oxaliplatin and to identify the molecular mechanisms associated with KO supplementation in a mouse model of colorectal cancer (CRC). METHODS Thirty-six male Balb/c mice were randomly divided into six groups. Five groups received standard chow diet supplemented with KO (150 g/kg)), corn oil (150 g/kg), KO combined with ½ dose of oxaliplatin (1.5 mg/kg body weight/3 times per week), corn oil combined with ½ dose of oxaliplatin (1.5 mg/kg body weight/3 times per week), or a full dose of oxaliplatin (3 mg/kg body weight/3 times per week). The control (sham) group received a standard chow diet. Treatments started three weeks before and continued for three weeks after orthotopic CRC induction. The number of metastases, tumour weight and volume were quantified ex-vivo. The expression of cytochrome c, cleaved caspase-9 and -3, DNA damage, PD-L1, PD-L2 and HSP-70 were determined. RESULTS A significant reductions in the weight and volume of tumours were observed in mice treated with KO and KO plus a ½ dose of oxaliplatin compared to the sham group, similar to oxaliplatin-treated mice. KO, and KO plus ½ dose of oxaliplatin significantly increased the expression of cytochrome c, cleaved caspase-9 and -3, and DNA damage and decreased expression of PD-L1, PD-L2 and HSP-70 in tumour tissues compared to the sham group. CONCLUSIONS The in vivo anti-cancer effects of KO are comparable with oxaliplatin. Thus, dietary KO supplementation has a great potential as a therapeutic/adjunctive agent for CRC treatment.
Collapse
Affiliation(s)
| | | | - Valentina Jovanovska
- Institute for Health and Sport, Victoria University, P.O. Box 14428, Melbourne, 8001, Australia
| | - Rodney Brain Luwor
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, P.O. Box 14428, Melbourne, 8001, Australia. .,Department of Medicine-Western Health, The University of Melbourne, Melbourne, Australia. .,Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Australia.
| | - Xiao Qun Su
- Institute for Health and Sport, Victoria University, P.O. Box 14428, Melbourne, 8001, Australia.
| |
Collapse
|
13
|
Sahyon HAE, Ramadan ENM, Althobaiti F, Mashaly MMA. Anti-proliferative effects of the combination of Sulfamethoxazole and Quercetin via caspase3 and NFkB gene regulation: an in vitro and in vivo study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:227-246. [PMID: 34994822 DOI: 10.1007/s00210-021-02174-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/27/2021] [Indexed: 01/30/2023]
Abstract
Combination therapy comprising natural polyphenols and anticancer drugs has been used to decrease the adverse effects and increase the effectiveness and antioxidant activities of the drugs. The antioxidant and anticancer effects of quercetin (Q), a nutritive polyphenol, have been observed both in vitro and in vivo. Likewise, the anticancer activity of sulfamethoxazole (S) has been demonstrated in vitro and in vivo. This study aimed to investigate the in vitro and in vivo anticancer effects of Q alone and in combination with S. The in vitro effects of S, Q, and S + Q on HCT-116, HepG2, MCF-7, and PC3 cell lines were examined. Additionally, the in vivo effects of these drugs were evaluated using Ehrlich ascites carcinoma (EAC) tumor-bearing mice. The in vitro data revealed the potent anticancer activity of S + Q through the induction of apoptosis and cell cycle arrest. The EAC-inoculated mice treated with S + Q presented with elevated SOD, GSH, CAT, and TAC levels and decreased malondialdehyde levels compared with the untreated EAC group, thus revealing the antioxidant and protective actions of S + Q against EAC cell invasion. Furthermore, the downregulation of NFkB and upregulation of the caspase3 gene in the EAC-inoculated mice treated with the S + Q indicated the induction of the apoptotic pathway and decrease in both cell proliferation and metastasis. In conclusion, the combination of S and Q might exert anticancer effects by inducing apoptosis and exhibiting selective toxicity against the cancer cells and thereby protecting the vital organs.
Collapse
Affiliation(s)
- Heba Abd Elghany Sahyon
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Elgiesh Street , Kafrelsheikh, 33516, Egypt.
| | - Eman N M Ramadan
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34518, Egypt
| | - Fayez Althobaiti
- Department of Biotechnology, Collage of Science, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Mohammad M A Mashaly
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34518, Egypt
| |
Collapse
|
14
|
Tummers B, Green DR. The evolution of regulated cell death pathways in animals and their evasion by pathogens. Physiol Rev 2022; 102:411-454. [PMID: 34898294 PMCID: PMC8676434 DOI: 10.1152/physrev.00002.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 09/01/2021] [Accepted: 09/01/2022] [Indexed: 12/21/2022] Open
Abstract
The coevolution of host-pathogen interactions underlies many human physiological traits associated with protection from or susceptibility to infections. Among the mechanisms that animals utilize to control infections are the regulated cell death pathways of pyroptosis, apoptosis, and necroptosis. Over the course of evolution these pathways have become intricate and complex, coevolving with microbes that infect animal hosts. Microbes, in turn, have evolved strategies to interfere with the pathways of regulated cell death to avoid eradication by the host. Here, we present an overview of the mechanisms of regulated cell death in Animalia and the strategies devised by pathogens to interfere with these processes. We review the molecular pathways of regulated cell death, their roles in infection, and how they are perturbed by viruses and bacteria, providing insights into the coevolution of host-pathogen interactions and cell death pathways.
Collapse
Affiliation(s)
- Bart Tummers
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
15
|
Dai H, Meng XW, Ye K, Jia J, Kaufmann SH. Therapeutics targeting BCL2 family proteins. MECHANISMS OF CELL DEATH AND OPPORTUNITIES FOR THERAPEUTIC DEVELOPMENT 2022:197-260. [DOI: 10.1016/b978-0-12-814208-0.00007-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Multi-Modal Biological Destruction by Cold Atmospheric Plasma: Capability and Mechanism. Biomedicines 2021; 9:biomedicines9091259. [PMID: 34572443 PMCID: PMC8465976 DOI: 10.3390/biomedicines9091259] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 01/07/2023] Open
Abstract
Cold atmospheric plasma (CAP) is a near-room-temperature, partially ionized gas composed of reactive neutral and charged species. CAP also generates physical factors, including ultraviolet (UV) radiation and thermal and electromagnetic (EM) effects. Studies over the past decade demonstrated that CAP could effectively induce death in a wide range of cell types, from mammalian to bacterial cells. Viruses can also be inactivated by a CAP treatment. The CAP-triggered cell-death types mainly include apoptosis, necrosis, and autophagy-associated cell death. Cell death and virus inactivation triggered by CAP are the foundation of the emerging medical applications of CAP, including cancer therapy, sterilization, and wound healing. Here, we systematically analyze the entire picture of multi-modal biological destruction by CAP treatment and their underlying mechanisms based on the latest discoveries particularly the physical effects on cancer cells.
Collapse
|
17
|
Reactive Oxygen Species (ROS) Regulates Different Types of Cell Death by Acting as a Rheostat. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9912436. [PMID: 34426760 PMCID: PMC8380163 DOI: 10.1155/2021/9912436] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/14/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are essential for cellular signaling and response to stress. The level of ROS and the type of ROS determine the ability of cells to undergo cell death. Furthermore, dysregulation of the antioxidant pathways is associated with many diseases. It has become apparent that cell death can occur through different mechanisms leading to the classifications of different types of cell death such as apoptosis, ferroptosis, and necroptosis. ROS play essential roles in all forms of cell death, but it is only now coming into focus that ROS control and determine the type of cell death that occurs in any given cell. Indeed, ROS may act as a rheostat allowing different cell death mechanisms to be engaged and crosstalk with different cell death types. In this review, we will describe the ROS regulatory pathways and how they control different types of cell death under normal and disease states. We will also propose how ROS could provide a mechanism of crosstalk between cell death mechanisms and act as a rheostat determining the type of cell death.
Collapse
|
18
|
Elsayed AM, Sherif NM, Hassan NS, Althobaiti F, Hanafy NAN, Sahyon HA. Novel quercetin encapsulated chitosan functionalized copper oxide nanoparticles as anti-breast cancer agent via regulating p53 in rat model. Int J Biol Macromol 2021; 185:134-152. [PMID: 34147524 DOI: 10.1016/j.ijbiomac.2021.06.085] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/01/2021] [Accepted: 06/12/2021] [Indexed: 01/18/2023]
Abstract
This study was designed to present a new quercetin encapsulated chitosan functionalized copper oxide nanoparticle (CuO-ChNPs-Q) and assessed its anti-breast cancer activity both in vitro and in vivo. The CuO-ChNPs-Q may act as anti-proliferating agent against DMBA-induced mammary carcinoma in female rats. The CuONPs was functionalized with chitosan then quercetin was conjugated with them producing CuO-ChNPs-Q, then characterized. The in vitro anti-proliferating activity of the CuO-ChNPs-Q was evaluated against three human cell line. Then, the anti-breast cancer effect of the CuO-ChNPs-Q was assessed against DMBA-induction compared to both CuONPs and Q in female rat model. The in vitro results proved the potent anticancer activity of the CuO-ChNPs-Q compared to CuONPs and quercetin. The in vivo data showed significant reduction in breast tumors of DMBA-induced rats treated with CuO-ChNPs-Q compared to CuONPs and Q. The CuO-ChNPs-Q treatment had induced apoptosis via increased p53 gene, arrested the cell-cycle, and increased both cytochrome c and caspase-3 levels leading to mammary carcinoma cell death. Also, the CuO-ChNPs-Q treatment had suppressed the PCNA gene which decreased the proliferation of the mammary carcinoma cells. In conclusion, the CuO-ChNPs-Q might be a promising chemotherapeutic agent for treatment of breast cancer with a minimal toxicity on vital organs.
Collapse
Affiliation(s)
- Awny M Elsayed
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Naglaa M Sherif
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Nahla S Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Fayez Althobaiti
- Department of Biotechnology, Collage of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Nemany A N Hanafy
- Nanomedicine group, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt.
| | - Heba A Sahyon
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt.
| |
Collapse
|
19
|
Yu ACY, Chern YJ, Zhang P, Pasiliao CC, Rahman M, Chang G, Ren J, Tai IT. Inhibition of nucleophosmin 1 suppresses colorectal cancer tumor growth of patient -derived xenografts via activation of p53 and inhibition of AKT. Cancer Biol Ther 2021; 22:112-123. [PMID: 33446037 DOI: 10.1080/15384047.2020.1839278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The nucleophosmin 1 (NPM1) protein is frequently overexpressed in various cancers compared to normal tissues and represents a potential biomarker for maliganancy. However, its role in colorectal cancer (CRC) is still not fully understood. In this report, we show that NPM1 levels in CRC correlate with prognosis and sensitivity to chemotherapy. NPM1 expression was found to be significantly increased in CRC tumors (P < .001) and was associated with poor overall 5-year survival (P < .05). For individuals with Stage IV disease, this represented a reduction in survival by 11 months (P < .01; HR = 0.38, CI [0.21, 0.69]. In vitro, we show that NPM1 gene silencing enhanced the chemosensitivity of CRC cells and that pharmacological inhibition of NPM1 by NSC348884 triggered the onset of programmed cell death. Our immunofluorescence microscopy and immunoblot analyses also revealed that blocking NPM1 function sensitized CRC cells to chemotherapy-induced apoptosis through a mechanism that involves proteins in the AKT pathway. Consistent with the in vitro data, our patient-derived CRC xenograft model showed that inhibition of NPM1 suppressed tumor growth and attenuated AKT signaling in vivo. Moreover, LY294002, an inhibitor of the PI3K/AKT pathway, restored the chemosensitivity of CRC cells expressing high levels of NPM1. The findings that NPM1's expression in CRC tissue correlates with prognosis and supports anti-apoptotic activity mediated by AKT signaling, further our understanding of the role of NPM1 in CRC.
Collapse
Affiliation(s)
- Angel C Y Yu
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Yi-Jye Chern
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Peter Zhang
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Clarissa C Pasiliao
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Mahbuba Rahman
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - George Chang
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Jianhua Ren
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Isabella T Tai
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
20
|
Li Z, Chen L, Chen C, Zhou Y, Hu D, Yang J, Chen Y, Zhuo W, Mao M, Zhang X, Xu L, Wang L, Zhou J. Targeting ferroptosis in breast cancer. Biomark Res 2020; 8:58. [PMID: 33292585 PMCID: PMC7643412 DOI: 10.1186/s40364-020-00230-3] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is a recently discovered distinct type of regulated cell death caused by the accumulation of lipid-based ROS. Metabolism and expression of specific genes affect the occurrence of ferroptosis, making it a promising therapeutic target to manage cancer. Here, we describe the current status of ferroptosis studies in breast cancer and trace the key regulators of ferroptosis back to previous studies. We also compare ferroptosis to common regulated cell death patterns and discuss the sensitivity to ferroptosis in different subtypes of breast cancer. We propose that viewing ferroptosis-related studies from a historical angle will accelerate the development of ferroptosis-based biomarkers and therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Zhaoqing Li
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), 2nd Affiliated Hospital, School of Medicine, Zhejiang University, 310009 Hangzhou, Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Lini Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Cong Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Yulu Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Dengdi Hu
- Cixi People’s Hospital Medical and Health Group, 315300 Ningbo, Zhejiang China
| | - Jingjing Yang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Yongxia Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Wenying Zhuo
- Cixi People’s Hospital Medical and Health Group, 315300 Ningbo, Zhejiang China
| | - Misha Mao
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Xun Zhang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Ling Xu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Linbo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| | - Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000 Zhejiang China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, 310000 Hangzhou, Zhejiang China
| |
Collapse
|
21
|
The role of KATP channel blockade and activation in the protection against neurodegeneration in the rotenone model of Parkinson's disease. Life Sci 2020; 257:118070. [DOI: 10.1016/j.lfs.2020.118070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/25/2020] [Accepted: 07/06/2020] [Indexed: 02/08/2023]
|
22
|
Vigneswara V, Ahmed Z. The Role of Caspase-2 in Regulating Cell Fate. Cells 2020; 9:cells9051259. [PMID: 32438737 PMCID: PMC7290664 DOI: 10.3390/cells9051259] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Caspase-2 is the most evolutionarily conserved member of the mammalian caspase family and has been implicated in both apoptotic and non-apoptotic signaling pathways, including tumor suppression, cell cycle regulation, and DNA repair. A myriad of signaling molecules is associated with the tight regulation of caspase-2 to mediate multiple cellular processes far beyond apoptotic cell death. This review provides a comprehensive overview of the literature pertaining to possible sophisticated molecular mechanisms underlying the multifaceted process of caspase-2 activation and to highlight its interplay between factors that promote or suppress apoptosis in a complicated regulatory network that determines the fate of a cell from its birth and throughout its life.
Collapse
|
23
|
The Effects of Houttuynia cordata Thunb and Piper ribesioides Wall Extracts on Breast Carcinoma Cell Proliferation, Migration, Invasion and Apoptosis. Molecules 2020; 25:molecules25051196. [PMID: 32155880 PMCID: PMC7179460 DOI: 10.3390/molecules25051196] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022] Open
Abstract
Houttuynia cordata Thunb. (HCT) and Piper ribesioides Wall. (PR) are common herbs that are widely distributed throughout East Asia and possess various biological properties including anti-cancer effects. However, in breast cancer, their mechanisms responsible for anti-carcinogenic effects have not been clarified yet. In this study, the inhibitory effects of HCT and PR ethanolic extracts on breast cancer cell proliferation, migration, invasion and apoptosis were examined. In MCF-7 and MDA-MB-231 cells, HCT and PR extracts at low concentrations can inhibit colony formation and induce G1 cell cycle arrest by downregulating cyclinD1 and CDK4 expression. Additionally, HCT and PR extracts also decreased the migration and invasion of both breast cancer cell lines through inhibition of MMP-2 and MMP-9 secretion. Moreover, the induction of apoptosis was observed in breast cancer cells treated with high concentrations of HCT and PR extracts. Not only stimulated caspases activity, but HCT and PR extracts also upregulated the expression of caspases and pro-apoptotic Bcl-2 family proteins in breast cancer cells. Altogether, these findings provide the rationale to further investigate the potential actions of HCT and PR extracts against breast cancer in vivo.
Collapse
|
24
|
Deng H, Ma J, Liu Y, He P, Dong W. Combining α-Hederin with cisplatin increases the apoptosis of gastric cancer in vivo and in vitro via mitochondrial related apoptosis pathway. Biomed Pharmacother 2019; 120:109477. [DOI: 10.1016/j.biopha.2019.109477] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023] Open
|
25
|
Current overview on the clinical update of Bcl-2 anti-apoptotic inhibitors for cancer therapy. Eur J Pharmacol 2019; 862:172655. [PMID: 31494078 DOI: 10.1016/j.ejphar.2019.172655] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 08/25/2019] [Accepted: 09/04/2019] [Indexed: 12/27/2022]
Abstract
Apoptosis is one of the major mechanisms exhibited in response to cell death and induction of apoptosis in tumour cells signifies a potential target for cancer therapy. Bcl-2 family proteins play a key role in regulation of the apoptotic pathway. Bcl-2 overexpression is commonly associated with various cancers including breast cancer, prostate cancer, B-cell lymphomas and colorectal adenocarcinomas etc. Thus, Bcl-2 is a novel anti-cancer target attracting medicinal chemists across the globe. Research investigations underlying Bcl-2 target have resulted in the generation of small molecule inhibitors, named as 'BH3-mimetics' (Bcl-2 homology 3 mimetics). These drugs display binding to pro-survival Bcl-2 proteins resulting in actuation of apoptosis of cancer cells. The first BH3 mimetics discovered as an outcome of structure-based drug design and Nuclear Magnetic Resonance (NMR)-based screening was ABT-263, an N-acylsulfonamide analogue. Thrombocytopenia a major dose-limiting toxicity, associated with ABT-263 had provoked the invention of a highly selective Bcl-2 inhibitor venetoclax. Several Bcl-2 inhibitors as small molecules are under clinical development and the results indicated that these molecules alone or in combination could be of potential application in cancer therapy. This review summarizes an up to date knowledge of the available small molecule inhibitors, their discovery, synthesis, current clinical and pre-clinical status.
Collapse
|
26
|
Yao W, Lin Z, Wang G, Li S, Chen B, Sui Y, Huang J, Liu Q, Shi P, Lin X, Liu Q, Yao H. Delicaflavone induces apoptosis via mitochondrial pathway accompanying G2/M cycle arrest and inhibition of MAPK signaling cascades in cervical cancer HeLa cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152973. [PMID: 31177019 DOI: 10.1016/j.phymed.2019.152973] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/22/2019] [Accepted: 05/25/2019] [Indexed: 05/22/2023]
Abstract
BACKGROUND Cervical cancer (CCa) represents the fourth most common cause of cancer-related death in women worldwide. CCa therapy is still a major clinical challenge worldwide. Finding and developing new anti-CCa chemotherapeutic drugs is a very significant issue. Delicaflavone is a rare biflavonoid from Selaginella doederleinii Hieron, which has shown strong anti-cancer activities in our preliminary screening. PURPOSE The present study aimed to investigate the apoptotic effect and mechanism of delicaflavone against CCa. METHODS In this study, the effect and potential mechanism of delicaflavone against CCa were investigated in vitro and in vivo by MTT assay, TEM, flow cytometry, western blot assay, qPCR assay, immunofluorescence assay and the mouse xenograft tumor model. RESULTS It was confirmed that delicaflavone inhibited the proliferation of human CCa HeLa cells, and induced morphological changes, G2/M phase arrest and apoptosis in a dose- and time-dependent manner. HeLa cells treated with delicaflavone showed the loss of mitochondrial membrane potential, release of Cytochrome c, activation of caspases, alteration of Bax/Bcl-2 balance, and the inhibition of MAPK signaling cascades. Furthermore, delicaflavone significantly decreased tumor growth in a dose-dependent manner without apparent side effects in a xenograft tumor model of HeLa cells. Immunohistochemistry analysis confirmed the up-regulation of Caspase-9, Caspase-3, Bax protein and down-regulation of Bcl-2 protein in the xenografts tumors, which was consistent with the results in vitro. CONCLUSION The results of the current study show that apoptosis is induced by the mitochondrial pathway accompanying with G2/M cycle arrest and inhibition of MAPK signaling cascades in human CCa HeLa cells, which can be used as a promising therapeutic drug for CCa.
Collapse
Affiliation(s)
- Wensong Yao
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou 350122, China
| | - Zhen Lin
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Gang Wang
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou 350122, China
| | - Shaoguang Li
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Bing Chen
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou 350122, China
| | - Yuxia Sui
- Department of Pharmacy, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jianyong Huang
- Department of Pharmaceutical, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Qicai Liu
- Department of Reproductive Medicine Centre, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Peiying Shi
- Department of Traditional Chinese Medicine Resource and Bee Products, Bee Science College, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xinhua Lin
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou 350122, China.
| | - Qicai Liu
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou 350122, China
| | - Hong Yao
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
27
|
Mitra S, Nguyen LN, Akter M, Park G, Choi EH, Kaushik NK. Impact of ROS Generated by Chemical, Physical, and Plasma Techniques on Cancer Attenuation. Cancers (Basel) 2019; 11:E1030. [PMID: 31336648 PMCID: PMC6678366 DOI: 10.3390/cancers11071030] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022] Open
Abstract
For the last few decades, while significant improvements have been achieved in cancer therapy, this family of diseases is still considered one of the deadliest threats to human health. Thus, there is an urgent need to find novel strategies in order to tackle this vital medical issue. One of the most pivotal causes of cancer initiation is the presence of reactive oxygen species (ROS) inside the body. Interestingly, on the other hand, high doses of ROS possess the capability to damage malignant cells. Moreover, several important intracellular mechanisms occur during the production of ROS. For these reasons, inducing ROS inside the biological system by utilizing external physical or chemical methods is a promising approach to inhibit the growth of cancer cells. Beside conventional technologies, cold atmospheric plasmas are now receiving much attention as an emerging therapeutic tool for cancer treatment due to their unique biophysical behavior, including the ability to generate considerable amounts of ROS. This review summarizes the important mechanisms of ROS generated by chemical, physical, and plasma approaches. We also emphasize the biological effects and cancer inhibition capabilities of ROS.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Linh Nhat Nguyen
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Mahmuda Akter
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Gyungsoon Park
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea.
| |
Collapse
|
28
|
Reid D, Mattos C. Targeting Cancer from a Structural Biology Perspective. UNRAVELLING CANCER SIGNALING PATHWAYS: A MULTIDISCIPLINARY APPROACH 2019:295-320. [DOI: 10.1007/978-981-32-9816-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
29
|
Chen Q, Zheng L, Mao Q, Liu J, Wang H, Jia D, Chen H, Wu W, Wei T. Fibrillar structures induced by a plant reovirus target mitochondria to activate typical apoptotic response and promote viral infection in insect vectors. PLoS Pathog 2019; 15:e1007510. [PMID: 30653614 PMCID: PMC6353215 DOI: 10.1371/journal.ppat.1007510] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 01/30/2019] [Accepted: 12/07/2018] [Indexed: 11/19/2022] Open
Abstract
Numerous plant viruses that cause significant agricultural problems are persistently transmitted by insect vectors. We wanted to see if apoptosis was involved in viral infection process in the vector. We found that a plant reovirus (rice gall dwarf virus, RGDV) induced typical apoptotic response during viral replication in the leafhopper vector and cultured vector cells, as demonstrated by mitochondrial degeneration and membrane potential decrease. Fibrillar structures formed by nonstructural protein Pns11 of RGDV targeted the outer membrane of mitochondria, likely by interaction with an apoptosis-related mitochondrial protein in virus-infected leafhopper cells or nonvector insect cells. Such association of virus-induced fibrillar structures with mitochondria clearly led to mitochondrial degeneration and membrane potential decrease, suggesting that RGDV Pns11 was the inducer of apoptotic response in insect vectors. A caspase inhibitor treatment and knockdown of caspase gene expression using RNA interference each reduced apoptosis and viral accumulation, while the knockdown of gene expression for the inhibitor of apoptosis protein improved apoptosis and viral accumulation. Thus, RGDV exploited caspase-dependent apoptotic response to promote viral infection in insect vectors. For the first time, we directly confirmed that a nonstructural protein encoded by a persistent plant virus can induce the typical apoptotic response to benefit viral transmission by insect vectors.
Collapse
Affiliation(s)
- Qian Chen
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Limin Zheng
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
- Key Laboratory of Pest Management of Horticultural Crop of Hunan Province, Hunan Plant Protection Institute, Hunan Academy of Agricultural Science, Changsha, PR China
| | - Qianzhuo Mao
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Jiejie Liu
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Haitao Wang
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Dongsheng Jia
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Hongyan Chen
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Wei Wu
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Taiyun Wei
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| |
Collapse
|
30
|
Mahgoub AM, Mahmoud MG, Selim MS, EL Awady ME. Exopolysaccharide from Marine Bacillus velezensis MHM3 Induces Apoptosis of Human Breast Cancer MCF-7 Cells through a Mitochondrial Pathway. Asian Pac J Cancer Prev 2018; 19:1957-1963. [PMID: 30051679 PMCID: PMC6165642 DOI: 10.22034/apjcp.2018.19.7.1957] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/15/2018] [Indexed: 11/27/2022] Open
Abstract
Objective: The production of new natural pharmaceutical agents that increase the efficiency of chemotherapy without affecting the normal cells is the goal of all researchers. Therefore, the present study expects to evaluate the antioxidant and anticancer studies against MCF-7 cell lines of EPS produced by novel Egyptian marine bacterial strain. Methods: Marine bacterium was isolated, purified and identified by 16S rRNA gene amplification and sequence analyses. MHMEPS (the produced EPS) was analyzed by Fourier Transform Infra-red (FTIR), monosugars identification by HPLC, molecular weight estimation and sulfur content were determined. While, in-vitro antioxidants characters was determined using various methods and anticancer studies against MCF-7 cell lines. Results: Bacillus velezensis MHM3 produced 5.8 g/L of MHMEPS. The chemical analysis of MHMEPS showed 24% uronic acid and 18.19% sulfate and monosugars glucuronic acid, glucose, fructose and rhamnose with molar ratio of 4.00: 2.00: 1.00: 0.13, correspondingly, with an overall weight average molecular weight Mw of 1.145×104 g/mol and the number average of molecular weights Mn of 5.155 ×103 g/mol. The FTIR analysis and periodate oxidation indicate the existence of β-(1–4) linkage acidic polysaccharide. MHMEPS showed antioxidant scavenging activity against DPPH•, H2O2 and Metal chelating activity, respectively. So, reducing power method give high activity at 500 μg/ml. MHMEPS hinder the proliferation of MCF-7 cells at 5-80 μg/ml compared to the control group. Moreover, induced apoptosis was associated with activation of caspase-3. Also increased cytochrome C levels significantly in a dose-dependent manner compared with the control. The Caspase-3 activity was raised in MHMEPS treated MCF-7 cells compared with the control (p<0.05) in a dose-dependent manner. Therefore, the result of DNA fragmentation was confirmed by DNA ladder assay. We presume that MHMEPS has high potential at its low concentration, as a novel restorative agent for the treatment of MCF-7 cells, with no cytotoxicity against normal cells.
Collapse
Affiliation(s)
- Ahmed M Mahgoub
- Molecular Drug Evaluation Department, National Organization for Drug Control and Research, Giza, Egypt.
| | | | | | | |
Collapse
|
31
|
Shutting Down Acute Myeloid Leukemia and Myelodysplastic Syndrome with BCL-2 Family Protein Inhibition. Curr Hematol Malig Rep 2018; 13:256-264. [DOI: 10.1007/s11899-018-0464-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
32
|
Chen Q, Li KT, Tian S, Yu TH, Yu LH, Lin HD, Bai DQ. Photodynamic Therapy Mediated by Aloe-Emodin Inhibited Angiogenesis and Cell Metastasis Through Activating MAPK Signaling Pathway on HUVECs. Technol Cancer Res Treat 2018; 17:1533033818785512. [PMID: 30068242 PMCID: PMC6071160 DOI: 10.1177/1533033818785512] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 03/28/2018] [Accepted: 05/31/2018] [Indexed: 01/23/2023] Open
Abstract
Photodynamic therapy is a clinically used, minimally invasive therapeutic procedure that involves the application of photosensitizers which can locate in target cells and so be irradiated at a corresponding wavelength. Laser light irradiation activation of photosensitizers generates free reactive oxygen species, which induces selective cytotoxic activity in target cells. Within recent years, aloe-emodin as a photosensitizer has been successfully applied in photodynamic therapy applications. Angiogenesis plays an important role in tumor growth and metastasis; thus, the development of a novel target treatment for angiogenesis is essential in order to improve treatment therapeutics for cancer treatment. An essential step in angiogenesis involves the formation of tube-like structures during matrix degradation, rearrangement, and apoptosis of endothelial cells. In the present study, we investigated the mechanisms of photocytotoxicity induced by aloe-emodin in human umbilical vein endothelial cells. Analysis of cell proliferation results noted a significant decrease in cultured cells which received various concentrations of aloe-emodin and photodynamic therapy-induced light doses. Additionally, mitochondrial mechanisms of apoptotic cell death were observed in aloe-emodin photodynamic therapy-treated cells, as tube formation assays noted angiogenesis suppression after treatment. The capacity of migration and invasion of human umbilical vein endothelial cells was measured using the transwell assay and demonstrated that aloe-emodin photodynamic therapy significantly inhibited the migration and invasion of human umbilical vein endothelial cells. The expression of p38, extracellular signal-regulated kinase, the c-Jun N-terminal kinases, and vascular endothelial growth factor suggested that the cellular metastasis was related to mitogen-activated protein kinase signal pathway. Furthermore, disorganization of F action cytoskeleton components was observed after aloe-emodin photodynamic therapy. Overall, the findings from this study suggest that aloe-emodin photodynamic therapy inhibited angiogenesis and cellular metastasis in human umbilical vein endothelial cells by activating the mitogen-activated protein kinase apoptotic signaling cell death pathway.
Collapse
Affiliation(s)
- Qing Chen
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing
Medical University, Chongqing, China
| | - Kai-Ting Li
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing
Medical University, Chongqing, China
| | - Si Tian
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing
Medical University, Chongqing, China
| | - Ting-He Yu
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of
Chongqing Medical University, Chongqing, China
| | - Le-Hu Yu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing
Medical University, Chongqing, China
| | - Hai-Dan Lin
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing
Medical University, Chongqing, China
| | - Ding-Qun Bai
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing
Medical University, Chongqing, China
| |
Collapse
|
33
|
Yan D, Sherman JH, Keidar M. Cold atmospheric plasma, a novel promising anti-cancer treatment modality. Oncotarget 2017; 8:15977-15995. [PMID: 27845910 PMCID: PMC5362540 DOI: 10.18632/oncotarget.13304] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/29/2016] [Indexed: 01/01/2023] Open
Abstract
Over the past decade, cold atmospheric plasma (CAP), a near room temperature ionized gas has shown its promising application in cancer therapy. Two CAP devices, namely dielectric barrier discharge and plasma jet, show significantly anti-cancer capacity over dozens of cancer cell lines in vitro and several subcutaneous xenograft tumors in vivo. In contrast to conventional anti-cancer approaches and drugs, CAP is a selective anti-cancer treatment modality. Thus far establishing the chemical and molecular mechanism of the anti-cancer capacity of CAP is far from complete. In this review, we provide a comprehensive introduction of the basics of CAP, state of the art research in this field, the primary challenges, and future directions to cancer biologists.
Collapse
Affiliation(s)
- Dayun Yan
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA
| | - Jonathan H Sherman
- Department of Neurological Surgery, The George Washington University,Washington, DC, USA
| | - Michael Keidar
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA
| |
Collapse
|
34
|
Dagbay KB, Hill ME, Barrett E, Hardy JA. Tumor-Associated Mutations in Caspase-6 Negatively Impact Catalytic Efficiency. Biochemistry 2017; 56:4568-4577. [PMID: 28726391 DOI: 10.1021/acs.biochem.7b00357] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Unregulated, particularly suppressed programmed cell death is one of the distinguishing features of many cancer cells. The cysteine protease caspase-6, one of the executioners of apoptotic cell death, plays a crucial role in regulation of apoptosis. Several somatic mutations in the CASP6 gene in tumor tissues have been reported. This work explores the effect of CASP6 tumor-associated mutations on the catalytic efficiency and structure of caspase-6. In general, these mutations showed decreased overall rates of catalytic turnover. Mutations within 8 Å of the substrate-binding pocket of caspase-6 were found to be the most catalytically deactivating. Notably, the R259H substitution decreased activity by 457-fold. This substitution disrupts the cation-π stacking interaction between Arg-259 and Trp-227, which is indispensable for proper assembly of the substrate-binding loops in caspase-6. Sequence conservation analysis at the homologous position across the caspase family suggests a role for this cation-π stacking in the catalytic function of caspases generally. These data suggest that caspase-6 deactivating mutations may contribute to multifactorial carcinogenic transformations.
Collapse
Affiliation(s)
- Kevin B Dagbay
- Department of Chemistry, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Maureen E Hill
- Department of Chemistry, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Elizabeth Barrett
- Department of Chemistry, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Jeanne A Hardy
- Department of Chemistry, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| |
Collapse
|
35
|
Bartel A, Göhler A, Hopf V, Breitbach K. Caspase-6 mediates resistance against Burkholderia pseudomallei infection and influences the expression of detrimental cytokines. PLoS One 2017; 12:e0180203. [PMID: 28686630 PMCID: PMC5501493 DOI: 10.1371/journal.pone.0180203] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/13/2017] [Indexed: 01/09/2023] Open
Abstract
Caspase-6 is a member of the executioner caspases and known to play a role in innate and adaptive immune processes. However, its role in infectious diseases has rarely been addressed yet. We here examined the impact of caspase-6 in an in vivo infection model using the Gram-negative rod Burkholderia pseudomallei, causing the infectious disease melioidosis that is endemic in tropical and subtropical areas around the world. Caspase-6-/- and C57BL/6 wild type mice were challenged with B. pseudomallei for comparing mortality, bacterial burden and inflammatory cytokine expression. Bone-marrow derived macrophages were used to analyse the bactericidal activity in absence of caspase-6. Caspase-6 deficiency was associated with higher mortality and bacterial burden in vivo after B. pseudomallei infection. The bactericidal activity of caspase-6-/- macrophages was impaired compared to wild type cells. Caspase-6-/- mice showed higher expression of the IL-1β gene, known to be detrimental in murine melioidosis. Expression of the IL-10 gene was also increased in caspase-6-/- mice as early as 6 hours after infection. Treatment with exogenous IL-10 rendered mice more susceptible against B. pseudomallei challenge. Thus, caspase-6 seems to play a crucial role for determining resistance against the causative agent of melioidosis. To our knowledge this is the first report showing that caspase-6 is crucial for mediating resistance in an in vivo infection model. Caspase-6 influences the expression of detrimental cytokines and therefore seems to be important for achieving a well-balanced immune response that contributes for an efficient elimination of the pathogen.
Collapse
Affiliation(s)
- Alexander Bartel
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - André Göhler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Verena Hopf
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Breitbach
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
- * E-mail:
| |
Collapse
|
36
|
Liu Z, Li G, Gou Y, Xiao D, Luo G, Saavedra JE, Liu J, Wang H. JS-K, a nitric oxide prodrug, induces DNA damage and apoptosis in HBV-positive hepatocellular carcinoma HepG2.2.15 cell. Biomed Pharmacother 2017; 92:989-997. [PMID: 28605880 DOI: 10.1016/j.biopha.2017.05.141] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 11/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most important cause of cancer-related death, and 85% of HCC is caused by chronic HBV infection, the prognosis of patients and the reduction of HBV DNA levels remain unsatisfactory. JS-K, a nitric oxide-releasing diazeniumdiolates, is effective against various tumors, but little is known on its effects on HBV positive HCC. We found that JS-K reduced the expression of HBsAg and HBeAg in HBV-positive HepG2.2.15 cells. This study aimed to further examine anti-tumor effects of JS-K on HepG2.2.15 cells. The MTT assay and colony forming assay were used to study the cell growth inhibition of JS-K; scratch assay and transwell assay were performed to detect cell migration. The cell cycle was detected by flow cytometry. The immunofluorescence, flow cytometry analysis, and western blot were used to study DNA damage and cell apoptosis. JS-K inhibited HepG2.2.15 cell growth in a dose-dependent manner, suppressed cell colony formation and migration, arrested cells gather in the G2 phase. JS-K (1-20μM) increased the expression of DNA damage-associated protein phosphorylation H2AX (γH2AX), phosphorylation of checkpoint kinase 1 (p-Chk1), phosphorylation of checkpoint kinase 2 (p-Chk2), ataxia-telangiectasia mutated (ATM), phosphorylation of ataxia-telangiectasia mutated rad3-related (p-ATR) and apoptotic-associated proteins cleaved caspase-3, cleaved caspase-7, cleaved poly ADP-ribose polymerase (cleaved PARP). The study demonstrated JS-K is effective against HBV-positive HepG2.2.15 cells, the mechanisms are not only related to inhibition of HBsAg and HBeAg secretion, but also related with induction of DNA damage and apoptosis. JS-K is a promising anti-cancer candidate against HBV-positive HCC.
Collapse
Affiliation(s)
- Zhengyun Liu
- Key Laboratory of infectious disease, Provincial Department of Education, Zunyi Medical College Guizhou, 563000 China; Research Center for Medicine and Biology, Zunyi Medical College, Guizhou, 563000 China; Key Lab for Basic Pharmacology of Ministry of Education, Zunyi Medical College, Guizhou, 563000 China
| | - Guangmin Li
- Department of emergency, Affiliated Hospital of Zunyi Medical College, China
| | - Ying Gou
- Key Laboratory of infectious disease, Provincial Department of Education, Zunyi Medical College Guizhou, 563000 China; Research Center for Medicine and Biology, Zunyi Medical College, Guizhou, 563000 China; Department of Microbiology, Zunyi Medical College, Guizhou, 563000 China
| | - Dongyan Xiao
- Key Laboratory of infectious disease, Provincial Department of Education, Zunyi Medical College Guizhou, 563000 China; Research Center for Medicine and Biology, Zunyi Medical College, Guizhou, 563000 China; Department of Microbiology, Zunyi Medical College, Guizhou, 563000 China
| | - Guo Luo
- Key Laboratory of infectious disease, Provincial Department of Education, Zunyi Medical College Guizhou, 563000 China; Research Center for Medicine and Biology, Zunyi Medical College, Guizhou, 563000 China
| | | | - Jie Liu
- Key Lab for Basic Pharmacology of Ministry of Education, Zunyi Medical College, Guizhou, 563000 China
| | - Huan Wang
- Key Laboratory of infectious disease, Provincial Department of Education, Zunyi Medical College Guizhou, 563000 China; Research Center for Medicine and Biology, Zunyi Medical College, Guizhou, 563000 China; Department of Microbiology, Zunyi Medical College, Guizhou, 563000 China.
| |
Collapse
|
37
|
Yu H, Guo P, Xie X, Wang Y, Chen G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J Cell Mol Med 2016; 21:648-657. [PMID: 27860262 PMCID: PMC5345622 DOI: 10.1111/jcmm.13008] [Citation(s) in RCA: 491] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 09/08/2016] [Indexed: 12/19/2022] Open
Abstract
Ferroptosis is a newly discovered type of cell death that differs from traditional apoptosis and necrosis and results from iron‐dependent lipid peroxide accumulation. Ferroptotic cell death is characterized by cytological changes, including cell volume shrinkage and increased mitochondrial membrane density. Ferroptosis can be induced by two classes of small‐molecule substances known as class 1 (system Xc− inhibitors) and class 2 ferroptosis inducers [glutathione peroxidase 4 (GPx4) inhibitors]. In addition to these small‐molecule substances, a number of drugs (e.g. sorafenib, artemisinin and its derivatives) can induce ferroptosis. Various factors, such as the mevalonate (MVA) and sulphur‐transfer pathways, play pivotal roles in the regulation of ferroptosis. Ferroptosis plays an unneglectable role in regulating the growth and proliferation of some types of tumour cells, such as lymphocytoma, ductal cell cancer of the pancreas, renal cell carcinoma (RCC) and hepatocellular carcinoma (HCC). Here, we will first introduce the discovery of and research pertaining to ferroptosis; then summarize the induction mechanisms and regulatory pathways of ferroptosis; and finally, further elucidate the roles of ferroptosis in human tumourous diseases.
Collapse
Affiliation(s)
- Haitao Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Pengyi Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaozai Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yi Wang
- Environmental and Public Health School of Wenzhou Medical University, Wenzhou, China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
38
|
Baumann KW, Baust JM, Snyder KK, Baust JG, Van Buskirk RG. Characterization of Pancreatic Cancer Cell Thermal Response to Heat Ablation or Cryoablation. Technol Cancer Res Treat 2016; 16:393-405. [PMID: 27340260 DOI: 10.1177/1533034616655658] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
One of the most lethal carcinomas is pancreatic cancer. As standard treatment using chemotherapy and radiation has shown limited success, thermal regimens (cryotherapy or heat ablation) are emerging as viable alternatives. Although promising, our understanding of pancreatic cancer response to thermal ablation remains limited. In this study, we investigated the thermal responses of 2 pancreatic cancer cell lines in an effort to identify the minimum lethal temperature needed for complete cell death to provide guidance for in vivo applications. PANC-1 and BxPC-3 were frozen (-10°C to -25°C) or heated (45°C-50°C) in single and repeated exposure regimes. Posttreatment survival and recovery were analyzed using alamarBlue assay over a 7-day interval. Modes of cell death were assessed using fluorescence microscopy (calcein acetoxymethyl ester/propidium iodide) and flow cytometry (YO-PRO-1/propidium iodide). Freezing to -10°C resulted in minimal cell death. Exposure to -15°C had a mild impact on PANC-1 survival (93%), whereas BxPC-3 was more severely damaged (33%). Exposure to -20°C caused a significant reduction in viability (PANC-1 = 23%; BxPC-3 = 2%) whereas -25°C yielded complete death. Double freezing exposure was more effective than single exposure. Repeat exposure to -15°C resulted in complete death of BxPC-3, whereas -20°C severely impacted PANC-1 (7%). Heating to 45°C resulted in minimum cell death. Exposure to 48°C yielded a slight increase in cell loss (PANC-1 = 85%; BxPC-3 = 98%). Exposure to 50°C caused a significant decline (PANC-1 = 70%; BxPC-3 = 9%) with continued deterioration to 0%. Double heating to 45°C resulted in similar effects observed in single exposures, whereas repeated 48°C resulted in significant increases in cell death (PANC-1 = 68%; BxPC-3 = 29%). In conclusion, we observed that pancreatic cancer cells were completely destroyed at temperatures <-25°C or >50°C using single thermal exposures. Repeated exposures resulted in increased cell death at less extreme temperatures. Our data suggest that thermal ablation strategies (heat or cryoablation) may represent a viable technique for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Kenneth W Baumann
- 1 Institute of Biomedical Technology, State University of New York, Binghamton, NY, USA.,2 Department of Biological Sciences, Binghamton University, Binghamton, NY, USA.,3 CPSI Biotech, Owego, NY, USA
| | - John M Baust
- 1 Institute of Biomedical Technology, State University of New York, Binghamton, NY, USA.,3 CPSI Biotech, Owego, NY, USA
| | - Kristi K Snyder
- 1 Institute of Biomedical Technology, State University of New York, Binghamton, NY, USA.,3 CPSI Biotech, Owego, NY, USA
| | - John G Baust
- 1 Institute of Biomedical Technology, State University of New York, Binghamton, NY, USA.,2 Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Robert G Van Buskirk
- 1 Institute of Biomedical Technology, State University of New York, Binghamton, NY, USA.,2 Department of Biological Sciences, Binghamton University, Binghamton, NY, USA.,3 CPSI Biotech, Owego, NY, USA
| |
Collapse
|
39
|
Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol 2016; 37:8471-86. [PMID: 27059734 DOI: 10.1007/s13277-016-5035-9] [Citation(s) in RCA: 408] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/28/2016] [Indexed: 12/12/2022] Open
Abstract
As much as the cellular viability is important for the living organisms, the elimination of unnecessary or damaged cells has the opposite necessity for the maintenance of homeostasis in tissues, organs and the whole organism. Apoptosis, a type of cell death mechanism, is controlled by the interactions between several molecules and responsible for the elimination of unwanted cells from the body. Apoptosis can be triggered by intrinsically or extrinsically through death signals from the outside of the cell. Any abnormality in apoptosis process can cause various types of diseases from cancer to auto-immune diseases. Different gene families such as caspases, inhibitor of apoptosis proteins, B cell lymphoma (Bcl)-2 family of genes, tumor necrosis factor (TNF) receptor gene superfamily, or p53 gene are involved and/or collaborate in the process of apoptosis. In this review, we discuss the basic features of apoptosis and have focused on the gene families playing critical roles, activation/inactivation mechanisms, upstream/downstream effectors, and signaling pathways in apoptosis on the basis of cancer studies. In addition, novel apoptotic players such as miRNAs and sphingolipid family members in various kind of cancer are discussed.
Collapse
|
40
|
Abstract
The role of caspase proteases in regulated processes such as apoptosis and inflammation has been studied for more than two decades, and the activation cascades are known in detail. Apoptotic caspases also are utilized in critical developmental processes, although it is not known how cells maintain the exquisite control over caspase activity in order to retain subthreshold levels required for a particular adaptive response while preventing entry into apoptosis. In addition to active site-directed inhibitors, caspase activity is modulated by post-translational modifications or metal binding to allosteric sites on the enzyme, which stabilize inactive states in the conformational ensemble. This review provides a comprehensive global view of the complex conformational landscape of caspases and mechanisms used to select states in the ensemble. The caspase structural database provides considerable detail on the active and inactive conformations in the ensemble, which provide the cell multiple opportunities to fine tune caspase activity. In contrast, the current database on caspase modifications is largely incomplete and thus provides only a low-resolution picture of global allosteric communications and their effects on the conformational landscape. In recent years, allosteric control has been utilized in the design of small drug compounds or other allosteric effectors to modulate caspase activity.
Collapse
Affiliation(s)
- A Clay Clark
- Department of Biology, University of Texas at Arlington , Arlington, Texas 76019, United States
| |
Collapse
|
41
|
Apoptotic Caspases in Promoting Cancer: Implications from Their Roles in Development and Tissue Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 930:89-112. [PMID: 27558818 DOI: 10.1007/978-3-319-39406-0_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoptosis, a major form of programmed cell death, is an important mechanism to remove extra or unwanted cells during development. In tissue homeostasis apoptosis also acts as a monitoring machinery to eliminate damaged cells in response to environmental stresses. During these processes, caspases, a group of proteases, have been well defined as key drivers of cell death. However, a wealth of evidence is emerging which supports the existence of many other non-apoptotic functions of these caspases, which are essential not only in proper organism development but also in tissue homeostasis and post-injury recovery. In particular, apoptotic caspases in stress-induced dying cells can activate mitogenic signals leading to proliferation of neighbouring cells, a phenomenon termed apoptosis-induced proliferation. Apparently, such non-apoptotic functions of caspases need to be controlled and restrained in a context-dependent manner during development to prevent their detrimental effects. Intriguingly, accumulating studies suggest that cancer cells are able to utilise these functions of caspases to their advantage to enable their survival, proliferation and metastasis in order to grow and progress. This book chapter will review non-apoptotic functions of the caspases in development and tissue homeostasis with focus on how these cellular processes can be hijacked by cancer cells and contribute to tumourigenesis.
Collapse
|
42
|
Apigenin Attenuates Melanoma Cell Migration by Inducing Anoikis through Integrin and Focal Adhesion Kinase Inhibition. Molecules 2015; 20:21157-66. [PMID: 26633318 PMCID: PMC6332386 DOI: 10.3390/molecules201219752] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 11/20/2015] [Accepted: 11/20/2015] [Indexed: 11/16/2022] Open
Abstract
Apigenin, a nonmutagenic flavonoid, has been found to have antitumor properties and is therefore particularly relevant for the development of chemotherapeutic agents for cancers. In this study, time- and dose-dependent cell viability and cytotoxicity were assessed to determine the effects of apigenin on A2058 and A375 melanoma cells. Melanoma cells were pretreated with different concentrations of apigenin and analyzed for morphological changes, anoikis induction, cell migration, and levels of proteins associated with apoptosis. Apigenin reduced integrin protein levels and inhibited the phosphorylation of focal adhesion kinase (FAK) and extracellular signal-regulated kinase (ERK1/2), which induce anoikis in human cutaneous melanoma cells. Apigenin exhibited dose-dependent inhibition of melanoma cell migration, unlike untreated controls. Furthermore, apigenin treatment increased apoptotic factors such as caspase-3 and cleaved poly(ADP-ribose) polymerase in a dose-dependent manner, demonstrating the metastasis of melanoma cells. Our results provide a new insight into the mechanisms by which apigenin prevents melanoma metastasis by sensitizing anoikis induced by the loss of integrin proteins in the FAK/ERK1/2 signaling pathway. These findings elucidate the related mechanisms and suggest the potential of apigenin in developing clinical treatment strategies against malignant melanoma.
Collapse
|
43
|
Zhou M, Li Y, Hu Q, Bai XC, Huang W, Yan C, Scheres SHW, Shi Y. Atomic structure of the apoptosome: mechanism of cytochrome c- and dATP-mediated activation of Apaf-1. Genes Dev 2015; 29:2349-61. [PMID: 26543158 PMCID: PMC4691890 DOI: 10.1101/gad.272278.115] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022]
Abstract
In this study, Zhou et al. report the first atomic structure of the mammalian apoptosome, determined at 3.8 Å resolution by cryo-electron microscopy. These findings provide novel insight into how CytC relieves the autoinhibition of Apaf-1 and how dATP triggers Apaf-1 oligomerization. The apoptotic protease-activating factor 1 (Apaf-1) controls the onset of many known forms of intrinsic apoptosis in mammals. Apaf-1 exists in normal cells as an autoinhibited monomer. Upon binding to cytochrome c and dATP, Apaf-1 oligomerizes into a heptameric complex known as the apoptosome, which recruits and activates cell-killing caspases. Here we present an atomic structure of an intact mammalian apoptosome at 3.8 Å resolution, determined by single-particle, cryo-electron microscopy (cryo-EM). Structural analysis, together with structure-guided biochemical characterization, uncovered how cytochrome c releases the autoinhibition of Apaf-1 through specific interactions with the WD40 repeats. Structural comparison with autoinhibited Apaf-1 revealed how dATP binding triggers a set of conformational changes that results in the formation of the apoptosome. Together, these results constitute the molecular mechanism of cytochrome c- and dATP-mediated activation of Apaf-1.
Collapse
Affiliation(s)
- Mengying Zhou
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yini Li
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qi Hu
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiao-Chen Bai
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom
| | - Weiyun Huang
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chuangye Yan
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Sjors H W Scheres
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom
| | - Yigong Shi
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
44
|
Hu Z, Zhou Q, Zhang C, Fan S, Cheng W, Zhao Y, Shao F, Wang HW, Sui SF, Chai J. Structural and biochemical basis for induced self-propagation of NLRC4. Science 2015; 350:399-404. [PMID: 26449475 DOI: 10.1126/science.aac5489] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/14/2015] [Indexed: 02/05/2023]
Abstract
Responding to stimuli, nucleotide-binding domain and leucine-rich repeat-containing proteins (NLRs) oligomerize into multiprotein complexes, termed inflammasomes, mediating innate immunity. Recognition of bacterial pathogens by NLR apoptosis inhibitory proteins (NAIPs) induces NLR family CARD domain-containing protein 4 (NLRC4) activation and formation of NAIP-NLRC4 inflammasomes. The wheel-like structure of a PrgJ-NAIP2-NLRC4 complex determined by cryogenic electron microscopy at 6.6 angstrom reveals that NLRC4 activation involves substantial structural reorganization that creates one oligomerization surface (catalytic surface). Once activated, NLRC4 uses this surface to catalyze the activation of an inactive NLRC4, self-propagating its active conformation to form the wheel-like architecture. NAIP proteins possess a catalytic surface matching the other oligomerization surface (receptor surface) of NLRC4 but not those of their own, ensuring that one NAIP is sufficient to initiate NLRC4 oligomerization.
Collapse
Affiliation(s)
- Zehan Hu
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qiang Zhou
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Chenlu Zhang
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shilong Fan
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Cheng
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yue Zhao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Feng Shao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Hong-Wei Wang
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Sen-Fang Sui
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Jijie Chai
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
45
|
Rajan S, Choi M, Baek K, Yoon HS. Bh3 induced conformational changes in Bcl-Xl revealed by crystal structure and comparative analysis. Proteins 2015; 83:1262-72. [PMID: 25907960 DOI: 10.1002/prot.24816] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 04/09/2015] [Accepted: 04/17/2015] [Indexed: 01/08/2023]
Abstract
Apoptosis or programmed cell death is a regulatory process in cells in response to stimuli perturbing physiological conditions. The Bcl-2 family of proteins plays an important role in regulating homeostasis during apoptosis. In the process, the molecular interactions among the three members of this family, the pro-apoptotic, anti-apoptotic and BH3-only proteins at the mitochondrial outer membrane define the fate of a cell. Here, we report the crystal structures of the human anti-apoptotic protein Bcl-XL in complex with BH3-only BID(BH3) and BIM(BH3) peptides determined at 2.0 Å and 1.5 Å resolution, respectively. The BH3 peptides bind to the canonical hydrophobic pocket in Bcl-XL and adopt an alpha helical conformation in the bound form. Despite a similar structural fold, a comparison with other BH3 complexes revealed structural differences due to their sequence variations. In the Bcl-XL-BID(BH3) complex we observed a large pocket, in comparison with other BH3 complexes, lined by residues from helices α1, α2, α3, and α5 located adjacent to the canonical hydrophobic pocket. These results suggest that there are differences in the mode of interactions by the BH3 peptides that may translate into functional differences in apoptotic regulation.
Collapse
Affiliation(s)
- Sreekanth Rajan
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Minjoo Choi
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Kwanghee Baek
- Department of Genetic Engineering, College of Life Sciences, Kyung Hee University, Yongin-Si, Gyeonggi-Do, 446-701, Republic of Korea
| | - Ho Sup Yoon
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.,Department of Genetic Engineering, College of Life Sciences, Kyung Hee University, Yongin-Si, Gyeonggi-Do, 446-701, Republic of Korea
| |
Collapse
|
46
|
Pang Y, Bai XC, Yan C, Hao Q, Chen Z, Wang JW, Scheres SHW, Shi Y. Structure of the apoptosome: mechanistic insights into activation of an initiator caspase from Drosophila. Genes Dev 2015; 29:277-87. [PMID: 25644603 PMCID: PMC4318144 DOI: 10.1101/gad.255877.114] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The autocatalytic activation of an initiator caspase, exemplified by caspase-9 in mammals or its ortholog, Dronc, in fruit flies, is facilitated by a multimeric adaptor complex known as the apoptosome. Pang et al. report two cryo-EM structures: the complete Dark apoptosome at an overall resolution of 4.0 Å and a complex between the Dark apoptosome and the CARD of Dronc at 4.1 Å resolution. The structural findings, together with structure-guided biochemical analyses, allow delineation of the molecular mechanisms for Dronc activation. Apoptosis is executed by a cascade of caspase activation. The autocatalytic activation of an initiator caspase, exemplified by caspase-9 in mammals or its ortholog, Dronc, in fruit flies, is facilitated by a multimeric adaptor complex known as the apoptosome. The underlying mechanism by which caspase-9 or Dronc is activated by the apoptosome remains unknown. Here we report the electron cryomicroscopic (cryo-EM) structure of the intact apoptosome from Drosophila melanogaster at 4.0 Å resolution. Analysis of the Drosophila apoptosome, which comprises 16 molecules of the Dark protein (Apaf-1 ortholog), reveals molecular determinants that support the assembly of the 2.5-MDa complex. In the absence of dATP or ATP, Dronc zymogen potently induces formation of the Dark apoptosome, within which Dronc is efficiently activated. At 4.1 Å resolution, the cryo-EM structure of the Dark apoptosome bound to the caspase recruitment domain (CARD) of Dronc (Dronc-CARD) reveals two stacked rings of Dronc-CARD that are sandwiched between two octameric rings of the Dark protein. The specific interactions between Dronc-CARD and both the CARD and the WD40 repeats of a nearby Dark protomer are indispensable for Dronc activation. These findings reveal important mechanistic insights into the activation of initiator caspase by the apoptosome.
Collapse
Affiliation(s)
- Yuxuan Pang
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiao-chen Bai
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Chuangye Yan
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qi Hao
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zheqin Chen
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jia-Wei Wang
- Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Sjors H W Scheres
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Yigong Shi
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China;
| |
Collapse
|
47
|
Targeting the Fas/FasL system in Rheumatoid Arthritis therapy: Promising or risky? Cytokine 2014; 75:228-33. [PMID: 25481649 DOI: 10.1016/j.cyto.2014.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 12/30/2022]
Abstract
Rheumatoid Arthritis (RA) is a chronic inflammatory disease affecting synovial joints. Tumor necrosis factor (TNF) α is a key component of RA pathogenesis and blocking this cytokine is the most common strategy to treat the disease. Though TNFα blockers are very efficient, one third of the RA patients are unresponsive or present side effects. Therefore, the development of novel therapeutic approaches is required. RA pathogenesis is characterized by the hyperplasia of the synovium, closely associated to the pseudo-tumoral expansion of fibroblast-like synoviocytes (FLS), which invade and destroy the joint structure. Hence, depletion of RA FLS has been proposed as an alternative therapeutic strategy. The TNF family member Fas ligand (FasL) was reported to trigger apoptosis in FLS of arthritic joints by binding to its receptor Fas and therefore suggested as a promising candidate for targeting the hyperplastic synovial tissue. However, this cytokine is pleiotropic and recent data from the literature indicate that Fas activation might have a disease-promoting role in RA by promoting cell proliferation. Therefore, a FasL-based therapy for RA requires careful evaluation before being applied. In this review we aim to overview what is known about the apoptotic and non-apoptotic effects of Fas/FasL system and discuss its relevance in RA.
Collapse
|
48
|
Moreira AC, Branco AF, Sampaio SF, Cunha-Oliveira T, Martins TR, Holy J, Oliveira PJ, Sardão VA. Mitochondrial apoptosis-inducing factor is involved in doxorubicin-induced toxicity on H9c2 cardiomyoblasts. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2468-78. [DOI: 10.1016/j.bbadis.2014.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 09/19/2014] [Accepted: 09/26/2014] [Indexed: 01/22/2023]
|
49
|
Wang XJ, Cao Q, Zhang Y, Su XD. Activation and regulation of caspase-6 and its role in neurodegenerative diseases. Annu Rev Pharmacol Toxicol 2014; 55:553-72. [PMID: 25340928 DOI: 10.1146/annurev-pharmtox-010814-124414] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Caspases, a family of cysteine proteases, are major mediators of apoptosis and inflammation. Caspase-6 is classified as an apoptotic effector, and it mediates nuclear shrinkage during apoptosis, but it possesses unique activation and regulation mechanisms that differ from those of other effector caspases. Furthermore, increasing evidence has shown that caspase-6 is highly involved in axon degeneration and neurodegenerative diseases, such as Huntington's disease and Alzheimer's disease. Cleavage at the caspase-6 site in mutated huntingtin protein is a prerequisite for the development of the characteristic behavioral and neuropathological features of Huntington's disease. Active caspase-6 is present in early stages of Alzheimer's disease, and caspase-6 activity is associated with the disease's pathological lesions. In this review, we discuss the evidence relevant to the role of caspase-6 in neurodegenerative diseases and summarize its activation and regulation mechanisms. In doing so, we provide new insight about potential therapeutic approaches that incorporate the modulation of caspase-6 function for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao-Jun Wang
- State Key Laboratory of Protein and Plant Gene Research and
| | | | | | | |
Collapse
|
50
|
Molecular determinants of caspase-9 activation by the Apaf-1 apoptosome. Proc Natl Acad Sci U S A 2014; 111:16254-61. [PMID: 25313070 DOI: 10.1073/pnas.1418000111] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Autocatalytic activation of an initiator caspase triggers the onset of apoptosis. In dying cells, caspase-9 activation is mediated by a multimeric adaptor complex known as the Apaf-1 apoptosome. The molecular mechanism by which caspase-9 is activated by the Apaf-1 apoptosome remains largely unknown. Here we demonstrate that the previously reported 1:1 interaction between Apaf-1 caspase recruitment domain (CARD) and caspase-9 CARD is insufficient for the activation of caspase-9. Rather, formation of a multimeric CARD:CARD assembly between Apaf-1 and caspase-9, which requires three types of distinct interfaces, underlies caspase-9 activation. Importantly, an additional surface area on the multimeric CARD assembly is essential for caspase-9 activation. Together, these findings reveal mechanistic insights into the activation of caspase-9 by the Apaf-1 apoptosome and support the induced conformation model for initiator caspase activation by adaptor complexes.
Collapse
|